Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
2.
Ther Innov Regul Sci ; 58(2): 273-284, 2024 03.
Article in English | MEDLINE | ID: mdl-38148473

ABSTRACT

BACKGROUND: In 2016, the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use updated its efficacy guideline for good clinical practice and introduced predefined quality tolerance limits (QTLs) as a quality control in clinical trials. QTLs are complementary to Quality by Design (QbD) principles (ICH-E8) and are one of the components of the risk-based clinical trial quality management system. METHODS: Currently the framework for QTLs process is well established, extensively describing the operational aspects of Defining, Monitoring and Reporting, but a single source of commonly used methods to establish QTLs and secondary limits is lacking. This paper will primarily focus on closing this gap and include applications of statistical process control and Bayesian methods on commonly used study level quality parameters such as premature treatment discontinuation, study discontinuation and significant protocol deviations as examples. CONCLUSIONS: Application of quality tolerance limits to parameters that correspond to critical to quality factors help identify systematic errors. Some situations pose special challenges to implementing QTLs and not all methods are optimal in every scenario. Early warning signals, in addition to QTL, are necessary to trigger actions to further minimize the possibility of an end-of-study excursion.


Subject(s)
Clinical Trials as Topic , Quality Control , Humans , Bayes Theorem
4.
J Nanobiotechnology ; 20(1): 378, 2022 Aug 13.
Article in English | MEDLINE | ID: mdl-35964037

ABSTRACT

BACKGROUND: Low-intensity pulsed ultrasound (LIPUS) has been reported to accelerate fracture healing, but the mechanism is unclear and its efficacy needs to be further optimized. Ultrasound in combination with functionalized microbubbles has been shown to induce local shear forces and controllable mechanical stress in cells, amplifying the mechanical effects of LIPUS. Nanoscale lipid bubbles (nanobubbles) have high stability and good biosafety. However, the effect of LIPUS combined with functionalized nanobubbles on osteogenesis has rarely been studied. RESULTS: In this study, we report cyclic arginine-glycine-aspartic acid-modified nanobubbles (cRGD-NBs), with a particle size of ~ 500 nm, able to actively target bone marrow mesenchymal stem cells (BMSCs) via integrin receptors. cRGD-NBs can act as nanomechanical force generators on the cell membrane, and further enhance the BMSCs osteogenesis and bone formation promoted by LIPUS. The polymerization of actin microfilaments and the mechanosensitive transient receptor potential melastatin 7 (TRPM7) ion channel play important roles in BMSCs osteogenesis promoted by LIPUS/cRGD-NBs. Moreover, the mutual regulation of TRPM7 and actin microfilaments promote the effect of LIPUS/cRGD-NBs. The extracellular Ca2 + influx, controlled partly by TRPM7, could participated in the effect of LIPUS/cRGD-NBs on BMSCs. CONCLUSIONS: The nanomechanical force generators cRGD-NBs could promote osteogenesis of BMSCs and bone formation induced by LIPUS, through regulation TRPM7, actin cytoskeleton, and intracellular calcium oscillations. This study provides new directions for optimizing the efficacy of LIPUS for fracture healing, and a theoretical basis for the further application and development of LIPUS in clinical practice.


Subject(s)
Mesenchymal Stem Cells , TRPM Cation Channels , Actin Cytoskeleton , Mesenchymal Stem Cells/metabolism , Osteogenesis , TRPM Cation Channels/metabolism , Ultrasonic Waves
5.
Stem Cell Res Ther ; 12(1): 269, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33957982

ABSTRACT

BACKGROUND: Sepsis is a systemic inflammatory response to a local severe infection that may lead to multiple organ failure and death. Previous studies have shown that 40-50% of patients with sepsis have diverse myocardial injuries and 70 to 90% mortality rates compared to 20% mortality in patients with sepsis without myocardial injury. Therefore, uncovering the mechanism of sepsis-induced myocardial injury and finding a target-based treatment are immensely important. OBJECTIVE: The present study elucidated the mechanism of sepsis-induced myocardial injury and examined the value of human umbilical cord mesenchymal stem cells (huMSCs) for protecting cardiac function in sepsis. METHODS: We used cecal ligation and puncture (CLP) to induce sepsis in mice and detect myocardial injury and cardiac function using serological markers and echocardiography. Cardiomyocyte apoptosis and heart tissue ultrastructure were detected using TdT-mediated dUTP Nick-End Labeling (TUNEL) and transmission electron microscopy (TEM), respectively. Fura-2 AM was used to monitor Ca2+ uptake and efflux in mitochondria. FQ-PCR and Western blotting detected expression of mitochondrial Ca2+ distribution regulators and PTEN-induced putative kinase 1 (PINK1). JC-1 was used to detect the mitochondrial membrane potential (Δψm) of cardiomyocytes. RESULTS: We found that expression of PINK1 decreased in mouse hearts during sepsis, which caused cardiomyocyte mitochondrial Ca2+ efflux disorder, mitochondrial calcium overload, and cardiomyocyte injury. In contrast, we found that exosomes isolated from huMSCs (huMSC-exo) carried Pink1 mRNA, which could be transferred to recipient cardiomyocytes to increase PINK1 expression. The reduction in cardiomyocyte mitochondrial calcium efflux was reversed, and cardiomyocytes recovered from injury. We confirmed the effect of the PINK1-PKA-NCLX axis on mitochondrial calcium homeostasis in cardiomyocytes during sepsis. CONCLUSION: The PINK1-PKA-NCLX axis plays an important role in mitochondrial calcium efflux in cardiomyocytes. Therefore, PINK1 may be a therapeutic target to protect cardiomyocyte mitochondria, and the application of huMSC-exo is a promising strategy against sepsis-induced heart dysfunction.


Subject(s)
Exosomes , Sepsis , Animals , Apoptosis , Calcium/metabolism , Humans , Mice , Mitochondria , Myocytes, Cardiac/metabolism , Protein Kinases/metabolism , Sepsis/metabolism
6.
Biochem Biophys Res Commun ; 515(1): 24-30, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31122700

ABSTRACT

Human umbilical cord-derived mesenchymal stromal cells (hUC-MSCs) in vitro expansion for long term may undergo epigenetic and genetic alterations that subsequently induce cellular senescence and associated growth inhibition. Increasing evidence implicated that aberrant histone acetylation modulates gene expression responsible for MSCs aging. Whether the dysregulation of p300 and its KAT activity is involved in the aging process of MSCs was still unexplored. In this study, we found a significant decrease of p300 but elevated p53/p21 levels in senescent hUC-MSCs at late-passage. Then we used two different approaches: (i) downregulation of p300 by siRNA and (ii) inhibition of the acetyltransferase(KAT) activity by C646 to determine the role of p300 in regulating MSCs senescence. We showed that inhibition of p300 induce premature senescence and decrease proliferation potential in hUC-MSCs. Moreover, upregulations of p53 and p21 expressions were confirmed in p300 knockdown and C646-treated hUC-MSCs. Taken together, these results suggest that p300 plays an important role in aging process of MSCs associated with activation of p53/p21 signaling pathway.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , E1A-Associated p300 Protein/deficiency , Mesenchymal Stem Cells/cytology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Umbilical Cord/cytology , Benzoates/pharmacology , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Nitrobenzenes , Pyrazoles/pharmacology , Pyrazolones , Signal Transduction/drug effects
7.
Cell Physiol Biochem ; 50(1): 41-51, 2018.
Article in English | MEDLINE | ID: mdl-30278461

ABSTRACT

BACKGROUND/AIMS: Maternally expressed gene 3 (MEG3) is an imprinted gene with maternal expression, which may function as a tumor suppressor by inhibiting angiogenesis. To identify the prognostic value of MEG3 in breast cancer, systematic analysis was performed in this study. METHODS: To evaluate gene alteration during breast carcinogenesis, we explored MEG3 expression using the Serial Analysis of Gene Expression Genie suite and Oncomine analysis. The prognostic roles of MEG3 in breast cancer were investigated using the PrognoScan database. The heat map and methylation status of MEG3 were determined using the UCSC Genome Browser. RESULTS: We found that MEG3 was more frequently downregulated in breast cancer than in normal tissues and this correlated with prognosis. However, estrogen receptor and progesterone receptor status were found to be positively correlated with MEG3 expression. Conversely, basal-like status, triple-negative breast cancer status, and Scarff Bloom & Richardson grade criterion were negatively correlated with MEG3 expression. Following data mining in multiple big data databases, we confirmed a positive correlation between MEG3 and heparan sulfate proteoglycan 2 (HSPG2) expression in breast cancer tissues. CONCLUSION: MEG3 could be adopted as a marker to predict the prognosis of breast cancer with HSPG2. However, large-scale and comprehensive research is needed to clarify our results.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Computational Biology/methods , RNA, Long Noncoding/metabolism , Adult , Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , DNA Methylation , Databases, Factual , Female , Gene Expression Regulation, Neoplastic , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Middle Aged , Prognosis , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , Survival Analysis , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
8.
J Cell Biochem ; 119(11): 8872-8886, 2018 11.
Article in English | MEDLINE | ID: mdl-30076626

ABSTRACT

Human mesenchymal stem cells (MSCs) are a heterogeneous subset of nonhematopoietic multipotent stromal stem cells and can differentiate into mesodermal lineage, such as adipocytes, osteocytes, and chondrocytes, as well as ectodermal and endodermal lineages. Human umbilical cord (UC) is one of the most promising sources of MSCs. However, the molecular and cellular characteristics of UC-derived MSCs (UC-MSCs) require extensive investigations, which are hampered by the limited lifespan and the diminished potency over passages. Here, we used the piggyBac transposon-based simian virus 40 T antigen (SV40T) immortalization system and effectively immortalized UC-MSCs, yielding the iUC-MSCs. A vast majority of the immortalized lines are positive for MSC markers but not for hematopoietic markers. The immortalization phenotype of the iUC-MSCs can be effectively reversed by flippase recombinase-induced the removal of SV40T antigen. While possessing long-term proliferation capability, the iUC-MSCs are not tumorigenic in vivo. Upon bone morphogenetic protein 9 (BMP9) stimulation, the iUC-MSC cells effectively differentiate into osteogenic, chondrogenic, and adipogenic lineages both in vitro and in vivo, which is indistinguishable from that of primary UC-MSCs, indicating that the immortalized UC-MSCs possess the characteristics similar to that of their primary counterparts and retain trilineage differentiation potential upon BMP9 stimulation. Therefore, the engineered iUC-MSCs should be a valuable alternative cell source for studying UC-MSC biology and their potential utilities in immunotherapies and regenerative medicine.


Subject(s)
Adipogenesis/physiology , Cell Differentiation/physiology , Growth Differentiation Factor 2/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Umbilical Cord/cytology , Analysis of Variance , Animals , Antigens, Polyomavirus Transforming/metabolism , Cell Culture Techniques/methods , Cell Proliferation , Chondrogenesis/physiology , Female , Genetic Vectors , HEK293 Cells , Humans , Infant, Newborn , Mice, Nude , Transposon Resolvases/metabolism
9.
Oncol Lett ; 15(5): 6203-6210, 2018 May.
Article in English | MEDLINE | ID: mdl-29616102

ABSTRACT

Reliable animal models are required for the in vivo study of the molecular mechanisms and effects of chemotherapeutic drugs in hepatocarcinoma. In vivo tracing techniques based on firefly luciferase (FLuc) may optimize the non-invasive monitoring of experimental animals. The present study established a murine Hepa1-6-FLuc cell line that stably expressed a retrovirus-delivered FLuc protein gene. The cell morphology, proliferation, migration and invasion ability of Hepa1-6-FLuc cells were the same as that of the Hepa1-6 cells, and thus is suitable to replace Hepa1-6 cells in the construction of hepatocarcinoma animal models. No differences in subcutaneous tumor mass and its pathomorphology from implanted Hepa1-6-FLuc cells were observed compared with Hepa1-6 control tumors. Bioluminescence imaging indicated that the Luc signal of the Hepa1-6-FLuc cells was consistently strengthened with increases in tumor mass; however, the Luc signal of Hepa1-6-AdFLuc became weaker and eventually disappeared during tumor development. Therefore, compared with the transient expression by adenovirus, stable expression of the FLuc gene in Hepa1-6 cells may better reflect cell proliferation and survival in vivo, and provide a reliable source for the establishment of hepatocarcinoma models.

10.
Biochem Biophys Res Commun ; 495(1): 78-85, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29050939

ABSTRACT

Mesenchymal stem cells (MSCs) have been widely studied as an attractive therapeutic agent for the treatment of tumors. However, the adverse effects of the tumor paracrine factors who affect MSCs are still unclear. In this study, we report for the first time that C6 glioma-conditioned medium (GCM) induces malignant transformation of MSCs. In contrast to MSCs, the transformed mesenchymal stem cells (TMCs) exhibited tumor cell characterizations in vitro and highly tumorigenic in vivo. Furthermore, GCM and recombinant S100B increased receptor for advanced glycation end products (RAGE) and its downstream Akt1, STAT3 genes expression as well as phosphorylation and transcriptional activation. Finally, blockage of S100B-RAGE interaction by RAGE inhibitor FPS-ZM1 attenuated GCM and S100B-induced Akt1, STAT3 activation, abolished its cell proliferation, migration and invasion actions. Together, these results suggest that the RAGE pathway may play a possible role in malignant transformation procedure of MSCs, and that this process may be mediated through S100B.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Glioma/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Culture Media, Conditioned , Gene Expression , Glioma/genetics , Glioma/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
11.
Oncotarget ; 8(32): 53581-53601, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28881833

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent progenitor cells that can undergo self-renewal and differentiate into multiple lineages. Osteogenic differentiation from MSCs is a well-orchestrated process and regulated by multiple signaling pathways. We previously demonstrated that BMP9 is one of the most potent osteogenic factors. However, molecular mechanism through which BMP9 governs osteoblastic differentiation remains to be fully understood. Increasing evidence indicates noncoding RNAs (ncRNAs) may play important regulatory roles in many physiological and/or pathologic processes. In this study, we investigate the role of lncRNA H19 in BMP9-regulated osteogenic differentiation of MSCs. We demonstrated that H19 was sharply upregulated at the early stage of BMP9 stimulation of MSCs, followed by a rapid decease and gradual return to basal level. This process was correlated with BMP9-induced expression of osteogenic markers. Interestingly, either constitutive H19 expression or silencing H19 expression in MSCs significantly impaired BMP9-induced osteogenic differentiation in vitro and in vivo, which was effectively rescued by the activation of Notch signaling. Either constitutive H19 expression or silencing H19 expression led to the increased expression of a group of miRNAs that are predicted to target Notch ligands and receptors. Thus, these results indicate that lncRNA H19 functions as an important mediator of BMP9 signaling by modulating Notch signaling-targeting miRNAs. Our findings suggest that the well-coordinated biphasic expression of lncRNA H19 may be essential in BMP9-induced osteogenic differentiation of MSCs, and that dysregulated H19 expression may impair normal osteogenesis, leading to pathogenic processes, such as bone tumor development.

12.
Genes Dis ; 4(2): 43-63, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28944281

ABSTRACT

With rapid advances in understanding molecular pathogenesis of human diseases in the era of genome sciences and systems biology, it is anticipated that increasing numbers of therapeutic genes or targets will become available for targeted therapies. Despite numerous setbacks, efficacious gene and/or cell-based therapies still hold the great promise to revolutionize the clinical management of human diseases. It is wildly recognized that poor gene delivery is the limiting factor for most in vivo gene therapies. There has been a long-lasting interest in using viral vectors, especially adenoviral vectors, to deliver therapeutic genes for the past two decades. Among all currently available viral vectors, adenovirus is the most efficient gene delivery system in a broad range of cell and tissue types. The applications of adenoviral vectors in gene delivery have greatly increased in number and efficiency since their initial development. In fact, among over 2,000 gene therapy clinical trials approved worldwide since 1989, a significant portion of the trials have utilized adenoviral vectors. This review aims to provide a comprehensive overview on the characteristics of adenoviral vectors, including adenoviral biology, approaches to engineering adenoviral vectors, and their applications in clinical and pre-clinical studies with an emphasis in the areas of cancer treatment, vaccination and regenerative medicine. Current challenges and future directions regarding the use of adenoviral vectors are also discussed. It is expected that the continued improvements in adenoviral vectors should provide great opportunities for cell and gene therapies to live up to its enormous potential in personalized medicine.

13.
J Cell Mol Med ; 21(11): 2782-2795, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28470873

ABSTRACT

The cranial suture complex is a heterogeneous tissue consisting of osteogenic progenitor cells and mesenchymal stem cells (MSCs) from bone marrow and suture mesenchyme. The fusion of cranial sutures is a highly coordinated and tightly regulated process during development. Craniosynostosis is a congenital malformation caused by premature fusion of cranial sutures. While the progenitor cells derived from the cranial suture complex should prove valuable for studying the molecular mechanisms underlying suture development and pathogenic premature suture fusion, primary human cranial suture progenitors (SuPs) have limited life span and gradually lose osteoblastic ability over passages. To overcome technical challenges in maintaining sufficient and long-term culture of SuPs for suture biology studies, we establish and characterize the reversibly immortalized human cranial suture progenitors (iSuPs). Using a reversible immortalization system expressing SV40 T flanked with FRT sites, we demonstrate that primary human suture progenitor cells derived from the patent sutures of craniosynostosis patients can be efficiently immortalized. The iSuPs maintain long-term proliferative activity, express most of the consensus MSC markers and can differentiate into osteogenic and adipogenic lineages upon BMP9 stimulation in vitro and in vivo. The removal of SV40 T antigen by FLP recombinase results in a decrease in cell proliferation and an increase in the endogenous osteogenic and adipogenic capability in the iSuPs. Therefore, the iSuPs should be a valuable resource to study suture development, intramembranous ossification and the pathogenesis of craniosynostosis, as well as to explore cranial bone tissue engineering.


Subject(s)
Cranial Sutures/metabolism , Craniosynostoses/genetics , Founder Effect , Growth Differentiation Factors/genetics , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation , Cell Line, Transformed , Cell Proliferation , Cranial Sutures/pathology , Craniosynostoses/metabolism , Craniosynostoses/pathology , Gene Expression , Growth Differentiation Factor 2 , Growth Differentiation Factors/metabolism , Humans , Infant , Male , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , Simian virus 40/genetics , Simian virus 40/metabolism , Transformation, Genetic
14.
Cell Physiol Biochem ; 41(5): 1905-1923, 2017.
Article in English | MEDLINE | ID: mdl-28384643

ABSTRACT

BACKGROUND/AIMS: Mesenchymal stem cells (MSCs) are multipotent progenitors that can differentiate into several lineages including bone. Successful bone formation requires osteogenesis and angiogenesis coupling of MSCs. Here, we investigate if simultaneous activation of BMP9 and Notch signaling yields effective osteogenesis-angiogenesis coupling in MSCs. METHODS: Recently-characterized immortalized mouse adipose-derived progenitors (iMADs) were used as MSC source. Transgenes BMP9, NICD and dnNotch1 were expressed by adenoviral vectors. Gene expression was determined by qPCR and immunohistochem¡stry. Osteogenic activity was assessed by in vitro assays and in vivo ectopic bone formation model. RESULTS: BMP9 upregulated expression of Notch receptors and ligands in iMADs. Constitutively-active form of Notch1 NICD1 enhanced BMP9-induced osteogenic differentiation both in vitro and in vivo, which was effectively inhibited by dominant-negative form of Notch1 dnNotch1. BMP9- and NICD1-transduced MSCs implanted with a biocompatible scaffold yielded highly mature bone with extensive vascularization. NICD1 enhanced BMP9-induced expression of key angiogenic regulators in iMADs and Vegfa in ectopic bone, which was blunted by dnNotch1. CONCLUSION: Notch signaling may play an important role in BMP9-induced osteogenesis and angiogenesis. It's conceivable that simultaneous activation of the BMP9 and Notch pathways should efficiently couple osteogenesis and angiogenesis of MSCs for successful bone tissue engineering.


Subject(s)
Growth Differentiation Factor 2/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Osteogenesis , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cell Line , Growth Differentiation Factor 2/genetics , Mesenchymal Stem Cells/cytology , Mice , Receptor, Notch1/genetics
15.
Oncotarget ; 8(16): 27105-27119, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28404920

ABSTRACT

The liver provides vital metabolic, exocrine and endocrine functions in the body as such pathological conditions of the liver lead to high morbidity and mortality. The liver is highly regenerative and contains facultative stem cells that become activated during injury to replicate to fully recover mass and function. Canonical Wnt/ß-catenin signaling plays an important role in regulating the proliferation and differentiation of liver progenitor cells during liver regeneration. However, possible roles of noncanonical Wnts in liver development and regeneration remain undefined. We previously established a reversibly-immortalized hepatic progenitor cell line (iHPx), which retains hepatic differentiation potential. Here, we analyze the expression pattern of the essential components of both canonical and noncanonical Wnt signaling pathways at different postnatal stages of mouse liver tissues and iHPx cells. We find that noncanonical Wnt4, Wnt5a, Wnt9b, Wnt10a and Wnt10b, are highly expressed concordantly with the high levels of canonical Wnts in late stages of liver tissues. Wnt5a, Wnt9b, Wnt10a and Wnt10b are able to antagonize Wnt3a-induced ß-catenin/TCF activity, reduce the stemness of iHPx cells, and promote hepatic differentiation of liver progenitors. Stem cell implantation assay demonstrates that Wnt5a, Wnt9b, Wnt10a and Wnt10b can inhibit cell proliferation and promote hepatic differentiation of the iHPx progenitor cells. Our results strongly suggest that noncanonical Wnts may play an important role in fine-tuning Wnt/ß-catenin functions during liver development and liver regeneration. Thus, understanding regulatory mechanisms governing proliferation and differentiation of liver progenitor cells may hold great promise to facilitate liver regeneration and/or progenitor cell-based therapies for liver diseases.


Subject(s)
Cell Differentiation , Cell Self Renewal , Liver/cytology , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Biomarkers , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Self Renewal/genetics , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Liver/physiology , Liver Regeneration/genetics , Male , Mice , Stem Cell Transplantation , Transcriptome
16.
ACS Appl Mater Interfaces ; 9(19): 15922-15932, 2017 May 17.
Article in English | MEDLINE | ID: mdl-28406027

ABSTRACT

Graphene-based materials are used in many fields but have found only limited applications in biomedicine, including bone tissue engineering. Here, we demonstrate that novel hybrid materials consisting of gelatin-derived graphene and silicate nanosheets of Laponite (GL) are biocompatible and promote osteogenic differentiation of mesenchymal stem cells (MSCs). Homogeneous cell attachment, long-term proliferation, and osteogenic differentiation of MSCs on a GL-scaffold were confirmed using optical microscopy and scanning electron microscopy. GL-powders made by pulverizing the GL-scaffold were shown to promote bone morphogenetic protein (BMP9)-induced osteogenic differentiation. GL-powders increased the alkaline phosphatase (ALP) activity in immortalized mouse embryonic fibroblasts but decreased the ALP activity in more-differentiated immortalized mouse adipose-derived cells. Note, however, that GL-powders promoted BMP9-induced calcium mineral deposits in both MSC lines, as assessed using qualitative and quantitative alizarin red assays. Furthermore, the expression of chondro-osteogenic regulator markers such as Runx2, Sox9, osteopontin, and osteocalcin was upregulated by the GL-powder, independent of BMP9 stimulation; although the powder synergistically upregulated the BMP9-induced Osterix expression, the adipogenic marker PPARγ was unaffected. Furthermore, in vivo stem cell implantation experiments demonstrated that GL-powder could significantly enhance the BMP9-induced ectopic bone formation from MSCs. Collectively, our results strongly suggest that the GL hybrid materials promote BMP9-induced osteogenic differentiation of MSCs and hold promise for the development of bone tissue engineering platforms.


Subject(s)
Mesenchymal Stem Cells , Animals , Cell Differentiation , Gelatin , Graphite , Growth Differentiation Factor 2 , Mice , Osteogenesis , Silicates
17.
Oncotarget ; 8(67): 111847-111865, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29340096

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells that can undergo self-renewal and differentiate into multi-lineages. Bone marrow stromal stem cells (BMSCs) represent one of the most commonly-used MSCs. In order to overcome the technical challenge of maintaining primary BMSCs in long-term culture, here we seek to establish reversibly immortalized mouse BMSCs (imBMSCs). By exploiting CRISPR/Cas9-based homology-directed-repair (HDR) mechanism, we target SV40T to mouse Rosa26 locus and efficiently immortalize mouse BMSCs (i.e., imBMSCs). We also immortalize BMSCs with retroviral vector SSR #41 and establish imBMSC41 as a control line. Both imBMSCs and imBMSC41 exhibit long-term proliferative capability although imBMSC41 cells have a higher proliferation rate. SV40T mRNA expression is 130% higher in imBMSC41 than that in imBMSCs. However, FLP expression leads to 86% reduction of SV40T expression in imBMSCs, compared with 63% in imBMSC41 cells. Quantitative genomic PCR analysis indicates that the average copy number of SV40T and hygromycin is 1.05 for imBMSCs and 2.07 for imBMSC41, respectively. Moreover, FLP expression removes 92% of SV40T in imBMSCs at the genome DNA level, compared with 58% of that in imBMSC41 cells, indicating CRISPR/Cas9 HDR-mediated immortalization of BMSCs can be more effectively reversed than that of retrovirus-mediated random integrations. Nonetheless, both imBMSCs and imBMSC41 lines express MSC markers and are highly responsive to BMP9-induced osteogenic, chondrogenic and adipogenic differentiation in vitro and in vivo. Thus, the engineered imBMSCs can be used as a promising alternative source of primary MSCs for basic and translational research in the fields of MSC biology and regenerative medicine.

18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(7): 958-62, 2016 Jul.
Article in Chinese | MEDLINE | ID: mdl-27363278

ABSTRACT

Objective To investigate the expressions of miR-125b and target gene Raf1 proto-oncogene serine/threonine protein kinase (RAF1) in peripheral blood mononuclear cells (PBMCs) of pediatric patients with pulmonary tuberculosis (PTB), and observe the regulation of miR-125b on macrophage apoptosis and activity. Methods PBMCs of patients with PTB and healthy children were collected and separated. Real-time fluorescence quantitative PCR was used to detect mRNA expression level of miR-125b and RAF1, and Western blotting was used to detect the protein level of RAF1. THP-1 macrophages were transfected into miR-125b mimic, negative control mimic (NC-mimic), miR-125b inhibitor and negative control inhibitor (NC-inhibitor), which were cultured for 48 hours. Western blotting was performed to observe the expression of RAF1 in THP-1 macrophages, annexin V-FITC/PI double staining combined with flow cytometry was used to test cell apoptosis, and CCK-8 assay was used to detect cell proliferation. Results The expression of miR-125b in PBMCs in pediatric patients with PTB was downregulated, and mRNA and protein levels of RAF1 were upregulated. When miR-125b was over-expressed in THP-1 macrophages, the expression of RAF1 was reduced to promote the apoptosis of macrophages and decrease cell activity; when the expression of miR-125b was inhibited in THP-1 macrophages, the expression of RAF1 was elevatedand the apoptosis of macrophages was inhibited, the cell activity was promoted. Conclusion In PBMCs of children with PTB, miR-125b level is low. Upregulation of miR-125b in THP-1 macrophages, the apoptosis of THP-1 macrophages is promoted and cell activity is inhibited.


Subject(s)
Apoptosis/genetics , Gene Expression , Macrophages/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-raf/genetics , Adolescent , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Child , Child, Preschool , Female , Flow Cytometry , Humans , Infant , Infant, Newborn , Leukocytes, Mononuclear/metabolism , Male , Proto-Oncogene Mas , Proto-Oncogene Proteins c-raf/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/metabolism
19.
Lab Invest ; 96(2): 116-36, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26618721

ABSTRACT

The canonical WNT/ß-catenin signaling pathway governs a myriad of biological processes underlying the development and maintenance of adult tissue homeostasis, including regulation of stem cell self-renewal, cell proliferation, differentiation, and apoptosis. WNTs are secreted lipid-modified glycoproteins that act as short-range ligands to activate receptor-mediated signaling pathways. The hallmark of the canonical pathway is the activation of ß-catenin-mediated transcriptional activity. Canonical WNTs control the ß-catenin dynamics as the cytoplasmic level of ß-catenin is tightly regulated via phosphorylation by the 'destruction complex', consisting of glycogen synthase kinase 3ß (GSK3ß), casein kinase 1α (CK1α), the scaffold protein AXIN, and the tumor suppressor adenomatous polyposis coli (APC). Aberrant regulation of this signaling cascade is associated with varieties of human diseases, especially cancers. Over the past decade, significant progress has been made in understanding the mechanisms of canonical WNT signaling. In this review, we focus on the current understanding of WNT signaling at the extracellular, cytoplasmic membrane, and intracellular/nuclear levels, including the emerging knowledge of cross-talk with other pathways. Recent progresses in developing novel WNT pathway-targeted therapies will also be reviewed. Thus, this review is intended to serve as a refresher of the current understanding about the physiologic and pathogenic roles of WNT/ß-catenin signaling pathway, and to outline potential therapeutic opportunities by targeting the canonical WNT pathway.


Subject(s)
Carcinogenesis , Neoplasms/drug therapy , Stem Cells , Wnt Proteins , Wnt Signaling Pathway , Animals , Drug Discovery , Humans , Mice
20.
Cell Physiol Biochem ; 37(6): 2375-92, 2015.
Article in English | MEDLINE | ID: mdl-26646427

ABSTRACT

BACKGROUND/AIMS: Although osteosarcoma (OS) is the most common primary malignancy of bone, its molecular pathogenesis remains to be fully understood. We previously found the calcium-binding protein S100A6 was expressed in ∼80% of the analyzed OS primary and/or metastatic tumor samples. Here, we investigate the role of S100A6 in OS growth and progression. METHODS: S100A6 expression was assessed by qPCR and Western blotting. Overexpression or knockdown of S100A6 was carried out to determine S100A6's effect on proliferation, cell cycle, apoptosis, tumor growth, and osteogenic differentiation. RESULTS: S100A6 expression was readily detected in human OS cell lines. Exogenous S100A6 expression promoted cell proliferation in vitro and tumor growth in an orthotopic xenograft model of human OS. S100A6 overexpression reduced the numbers of OS cells in G1 phase and increased viable cells under serum starvation condition. Conversely, silencing S100A6 expression induced the production of cleaved caspase 3, and increased early stage apoptosis. S100A6 knockdown increased osteogenic differentiation activity of mesenchymal stem cells, while S100A6 overexpression inhibited osteogenic differentiation. BMP9-induced bone formation was augmented by S100A6 knockdown. CONCLUSION: Our findings strongly suggest that S100A6 may promote OS cell proliferation and OS tumor growth at least in part by facilitating cell cycle progression, preventing apoptosis, and inhibiting osteogenic differentiation. Thus, it is conceivable that targeting S100A6 may be exploited as a novel anti-OS therapy.


Subject(s)
Cell Cycle Proteins/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Osteogenesis , Osteosarcoma/pathology , S100 Proteins/physiology , Animals , Cell Line, Tumor , Heterografts , Humans , Mice , S100 Calcium Binding Protein A6
SELECTION OF CITATIONS
SEARCH DETAIL
...